Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Am J Clin Nutr ; 118(1): 329-337, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37230178

RESUMO

On September 7 and 8, 2022, Healthy Environment and Endocrine Disruptors Strategies, an Environmental Health Sciences program, convened a scientific workshop of relevant stakeholders involved in obesity, toxicology, or obesogen research to review the state of the science regarding the role of obesogenic chemicals that might be contributing to the obesity pandemic. The workshop's objectives were to examine the evidence supporting the hypothesis that obesogens contribute to the etiology of human obesity; to discuss opportunities for improved understanding, acceptance, and dissemination of obesogens as contributors to the obesity pandemic; and to consider the need for future research and potential mitigation strategies. This report details the discussions, key areas of agreement, and future opportunities to prevent obesity. The attendees agreed that environmental obesogens are real, significant, and a contributor at some degree to weight gain at the individual level and to the global obesity and metabolic disease pandemic at a societal level; moreover, it is at least, in theory, remediable.


Assuntos
Disruptores Endócrinos , Exposição Ambiental , Humanos , Exposição Ambiental/efeitos adversos , Exposição Ambiental/prevenção & controle , Disruptores Endócrinos/toxicidade , Obesidade/epidemiologia , Obesidade/etiologia , Obesidade/metabolismo , Aumento de Peso , Pandemias
2.
Cell Rep ; 42(4): 112390, 2023 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-37053070

RESUMO

White adipose tissue (WAT) distribution is sex dependent. Adipocyte hyperplasia contributes to WAT distribution in mice driven by cues in the tissue microenvironment, with females displaying hyperplasia in subcutaneous and visceral WAT, while males and ovariectomized females have visceral WAT (VWAT)-specific hyperplasia. However, the mechanism underlying sex-specific hyperplasia remains elusive. Here, transcriptome analysis in female mice shows that high-fat diet (HFD) induces estrogen signaling in adipocyte precursor cells (APCs). Analysis of APCs throughout the estrous cycle demonstrates increased proliferation only when proestrus (high estrogen) coincides with the onset of HFD feeding. We further show that estrogen receptor α (ERα) is required for this proliferation and that estradiol treatment at the onset of HFD feeding is sufficient to drive it. This estrous influence on APC proliferation leads to increased obesity driven by adipocyte hyperplasia. These data indicate that estrogen drives ERα-dependent obesogenic adipocyte hyperplasia in females, exacerbating obesity and contributing to the differential fat distribution between the sexes.


Assuntos
Estradiol , Receptor alfa de Estrogênio , Masculino , Feminino , Animais , Camundongos , Hiperplasia/patologia , Estradiol/farmacologia , Adipócitos/patologia , Tecido Adiposo Branco , Obesidade/patologia , Estrogênios , Dieta Hiperlipídica/efeitos adversos , Camundongos Endogâmicos C57BL , Adipogenia
3.
iScience ; 26(1): 105750, 2023 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-36590177

RESUMO

Establishing metabolic programming begins during fetal and postnatal development, and early-life lipid exposures play a critical role during neonatal adipogenesis. We define how neonatal consumption of a low omega-6 to -3 fatty acid ratio (n6/n3 FA ratio) establishes FA oxidation in adipocyte precursor cells (APCs) before they become adipocytes. In vivo, APCs isolated from mouse pups exposed to the low n6/n3 FA ratio had superior FA oxidation capacity, elevated beige adipocyte mRNAs Ppargc1α, Ucp2, and Runx1, and increased nuclear receptor NR2F2 protein. In vitro, APC treatment with NR2F2 ligand-induced beige adipocyte mRNAs and increased mitochondrial potential but not mass. Single-cell RNA-sequencing analysis revealed low n6/n3 FA ratio yielded more mitochondrial-high APCs and linked APC NR2F2 levels with beige adipocyte signatures and FA oxidation. Establishing beige adipogenesis is of clinical relevance, because fat depots with energetically active, smaller, and more numerous adipocytes improve metabolism and delay metabolic dysfunction.

4.
Stem Cell Reports ; 17(5): 1081-1088, 2022 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-35487210

RESUMO

Fat distribution is sexually dimorphic and is associated with metabolic disease risk. It is unknown if prepubertal sex-hormone signaling influences adult fat distribution. Here, we show that karyotypically male androgen-insensitive mice exhibit pronounced subcutaneous adiposity compared with wild-type males and females. This subcutaneous adipose bias emerges prior to puberty and is not due to differences in adipocyte size or rates of adipogenesis between visceral and subcutaneous fat. Instead, we find that androgen-insensitive mice lack an adequate progenitor pool for normal visceral-fat expansion during development, thus increasing the subcutaneous-to-visceral-fat ratio. Obesogenic visceral-fat expansion is likewise inhibited in these mice, yet their metabolic health is similar to wild-type animals with comparable total fat mass. Taken together, these data show that adult fat distribution can be determined prior to the onset of puberty by the relative number of progenitors that seed nascent adipose depots.


Assuntos
Tecido Adiposo , Androgênios , Adipogenia/genética , Adiposidade , Animais , Feminino , Gordura Intra-Abdominal/metabolismo , Masculino , Camundongos
5.
J Mol Endocrinol ; 68(4): 179-194, 2022 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-35244608

RESUMO

Sex hormones play a pivotal role in physiology and disease. Estrogen, the female sex hormone, has been long implicated in having protective roles against obesity. However, the direct impact of estrogens in white adipose tissue (WAT) function and growth is not understood. Here, we show that the deletion of estrogen receptor alpha (ERα; Esr1) from adipocytes using Adipoq-credoes not affect adipose mass in male or female mice under normal or high-fat diet (HFD) conditions. However, loss of ERα in adipocyte precursor cells (APs) via Pdgfra-cre leads to exacerbated obesity upon HFD feeding in both male and female mice, with s.c. adipose (SWAT)-specific expansion in male mice. Further characterization of these mice revealed infertility and increased plasma levels of sex hormones, including estradiol in female mice and androgens in male mice. These findings compromise the study of estrogen signaling within the adipocyte lineage using the Pdgfra-crestrain. However, AP transplant studies demonstrate that the increased AP hyperplasia in male SWAT upon Pdgfra-cre-mediated ablation of ERα is not driven by AP-intrinsic mechanisms but is rather mediated by off-target effects. These data highlight the inherent difficulties in studying models that disrupt the intricate balance of sex hormones. Thus, better approaches are needed to study the cellular and molecular mechanisms of sex hormones in obesity and disease.


Assuntos
Adipócitos , Receptor alfa de Estrogênio , Tecido Adiposo Branco , Animais , Dieta Hiperlipídica/efeitos adversos , Receptor alfa de Estrogênio/genética , Estrogênios , Feminino , Hormônios Esteroides Gonadais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Obesidade/genética
6.
Mol Metab ; 44: 101141, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33307216

RESUMO

OBJECTIVE: Low testosterone in men (hypogonadism) is associated with obesity and type II diabetes. Testosterone replacement therapy has been shown to reverse these effects. However, the mechanisms by which testosterone regulates total fat mass, fat distribution, and metabolic health are unclear. In this study, we clarify the impact of hypogonadism on these parameters, as well as parse the role of testosterone from its downstream metabolites, dihydrotestosterone (DHT), and estradiol, in the regulation of depot-specific adipose tissue mass. METHODS: To achieve this objective, we utilized mouse models of male hypogonadism coupled with hormone replacement therapy, magnetic resonance imaging (MRI), glucose tolerance tests, flow cytometry, and immunohistochemical techniques. RESULTS: We observed that castrated mice develop increased fat mass, reduced muscle mass, and impaired glucose metabolism compared with gonadally intact males. Interestingly, obesity is further accelerated in castrated mice fed a high-fat diet, suggesting hypogonadism increases susceptibility to obesogenesis when dietary consumption of fat is elevated. By performing hormone replacement therapy in castrated mice, we show that testosterone impedes visceral and subcutaneous fat mass expansion. Testosterone-derived estradiol selectively blocks visceral fat growth, and DHT selectively blocks the growth of subcutaneous fat. These effects are mediated by depot-specific alterations in adipocyte size. We also show that high-fat diet-induced adipogenesis is elevated in castrated mice and that this can be rescued by androgen treatment. Obesogenic adipogenesis is also elevated in mice where androgen receptor activity is inhibited. CONCLUSIONS: These data indicate that hypogonadism impairs glucose metabolism and increases obesogenic fat mass expansion through adipocyte hypertrophy and adipogenesis. In addition, our findings highlight distinct roles for testosterone, DHT, and estradiol in the regulation of total fat mass and fat distribution and reveal that androgen signaling blocks obesogenic adipogenesis in vivo.


Assuntos
Distribuição da Gordura Corporal , Hipogonadismo/metabolismo , Obesidade/metabolismo , Testosterona/metabolismo , Adipócitos/metabolismo , Adipogenia/efeitos dos fármacos , Animais , Composição Corporal , Diabetes Mellitus Tipo 2/metabolismo , Dieta Hiperlipídica/efeitos adversos , Glucose/metabolismo , Gordura Intra-Abdominal/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Gordura Subcutânea/metabolismo
7.
Artigo em Inglês | MEDLINE | ID: mdl-32180758

RESUMO

The interest in bone marrow adiposity (BMA) has increased over the last decade due to its association with, and potential role, in a range of diseases (osteoporosis, diabetes, anorexia, cancer) as well as treatments (corticosteroid, radiation, chemotherapy, thiazolidinediones). However, to advance the field of BMA research, standardization of methods is desirable to increase comparability of study outcomes and foster collaboration. Therefore, at the 2017 annual BMA meeting, the International Bone Marrow Adiposity Society (BMAS) founded a working group to evaluate methodologies in BMA research. All BMAS members could volunteer to participate. The working group members, who are all active preclinical or clinical BMA researchers, searched the literature for articles investigating BMA and discussed the results during personal and telephone conferences. According to the consensus opinion, both based on the review of the literature and on expert opinion, we describe existing methodologies and discuss the challenges and future directions for (1) histomorphometry of bone marrow adipocytes, (2) ex vivo BMA imaging, (3) in vivo BMA imaging, (4) cell isolation, culture, differentiation and in vitro modulation of primary bone marrow adipocytes and bone marrow stromal cell precursors, (5) lineage tracing and in vivo BMA modulation, and (6) BMA biobanking. We identify as accepted standards in BMA research: manual histomorphometry and osmium tetroxide 3D contrast-enhanced µCT for ex vivo quantification, specific MRI sequences (WFI and H-MRS) for in vivo studies, and RT-qPCR with a minimal four gene panel or lipid-based assays for in vitro quantification of bone marrow adipogenesis. Emerging techniques are described which may soon come to complement or substitute these gold standards. Known confounding factors and minimal reporting standards are presented, and their use is encouraged to facilitate comparison across studies. In conclusion, specific BMA methodologies have been developed. However, important challenges remain. In particular, we advocate for the harmonization of methodologies, the precise reporting of known confounding factors, and the identification of methods to modulate BMA independently from other tissues. Wider use of existing animal models with impaired BMA production (e.g., Pfrt-/-, KitW/W-v) and development of specific BMA deletion models would be highly desirable for this purpose.


Assuntos
Adipogenia , Adiposidade , Medula Óssea/patologia , Obesidade/patologia , Projetos de Pesquisa/normas , Relatório de Pesquisa/normas , Animais , Guias como Assunto , Humanos , Agências Internacionais , Sociedades Científicas
8.
Endocr Rev ; 40(5): 1187-1206, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31127816

RESUMO

The presence of adipocytes in mammalian bone marrow (BM) has been recognized histologically for decades, yet, until recently, these cells have received little attention from the research community. Advancements in mouse transgenics and imaging methods, particularly in the last 10 years, have permitted more detailed examinations of marrow adipocytes than ever before and yielded data that show these cells are critical regulators of the BM microenvironment and whole-body metabolism. Indeed, marrow adipocytes are anatomically and functionally separate from brown, beige, and classic white adipocytes. Thus, areas of BM space populated by adipocytes can be considered distinct fat depots and are collectively referred to as marrow adipose tissue (MAT) in this review. In the proceeding text, we focus on the developmental origin and physiologic functions of MAT. We also discuss the signals that cause the accumulation and loss of marrow adipocytes and the ability of these cells to regulate other cell lineages in the BM. Last, we consider roles for MAT in human physiology and disease.


Assuntos
Adiposidade , Medula Óssea/metabolismo , Adipócitos , Animais , Medula Óssea/crescimento & desenvolvimento , Medula Óssea/fisiologia , Humanos , Transdução de Sinais
9.
Cell Rep ; 27(5): 1528-1540.e7, 2019 04 30.
Artigo em Inglês | MEDLINE | ID: mdl-31042478

RESUMO

Adipocyte progenitor cells (APCs) provide the reservoir of regenerative cells to produce new adipocytes, although their identity in humans remains elusive. Using FACS analysis, gene expression profiling, and metabolic and proteomic analyses, we identified three APC subtypes in human white adipose tissues. The APC subtypes are molecularly distinct but possess similar proliferative and adipogenic capacities. Adipocytes derived from APCs with high CD34 expression exhibit exceedingly high rates of lipid flux compared with APCs with low or no CD34 expression, while adipocytes produced from CD34- APCs display beige-like adipocyte properties and a unique endocrine profile. APCs were more abundant in gluteofemoral compared with abdominal subcutaneous and omental adipose tissues, and the distribution of APC subtypes varies between depots and in patients with type 2 diabetes. These findings provide a mechanistic explanation for the heterogeneity of human white adipose tissue and a potential basis for dysregulated adipocyte function in type 2 diabetes.


Assuntos
Gordura Abdominal/citologia , Adipócitos/metabolismo , Diabetes Mellitus Tipo 2/patologia , Células-Tronco Mesenquimais/metabolismo , Gordura Subcutânea/citologia , Gordura Abdominal/patologia , Adipócitos/classificação , Adipócitos/fisiologia , Adiposidade , Adulto , Animais , Antígenos CD34/genética , Antígenos CD34/metabolismo , Proliferação de Células , Células Cultivadas , Feminino , Humanos , Masculino , Células-Tronco Mesenquimais/classificação , Células-Tronco Mesenquimais/fisiologia , Camundongos , Camundongos SCID , Pessoa de Meia-Idade , Proteoma , Gordura Subcutânea/patologia , Transcriptoma
10.
Development ; 146(7)2019 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-30948523

RESUMO

Adipose tissue is composed of anatomically distinct depots that mediate several important aspects of energy homeostasis. The past two decades have witnessed increased research effort to elucidate the ontogenetic basis of adipose form and function. In this Review, we discuss advances in our understanding of adipose tissue development with particular emphasis on the embryonic patterning of depot-specific adipocyte lineages and adipocyte differentiation in vivo Micro-environmental cues and other factors that influence cell identity and cell behavior at various junctures in the adipocyte lineage hierarchy are also considered.


Assuntos
Adipócitos/citologia , Tecido Adiposo/citologia , Tecido Adiposo/metabolismo , Obesidade/metabolismo , Adipócitos/metabolismo , Adipogenia/genética , Adipogenia/fisiologia , Animais , Humanos
11.
Endocrinology ; 160(1): 205-219, 2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30445425

RESUMO

The increased hepatic gluconeogenesis in type 2 diabetes mellitus has often been ascribed to increased transcription of phosphoenolpyruvate carboxykinase 1, cystolic form (PEPCK1), although recent evidence has questioned this attribution. To assess the metabolic role of PEPCK1, we treated regular chow fed and high-fat fed (HFF) male Sprague-Dawley rats with a 2'-O-methoxyethyl chimeric antisense oligonucleotide (ASO) against PEPCK1 and compared them with control ASO-treated rats. PEPCK1 ASO effectively decreased PEPCK1 expression in the liver and white adipose tissue. In chow fed rats, PEPCK1 ASO did not alter adiposity, plasma glucose, or insulin. In contrast, PEPCK1 ASO decreased the white adipose tissue mass in HFF rats but without altering basal rates of lipolysis, de novo lipogenesis, or glyceroneogenesis in vivo. Despite the protection from adiposity, hepatic insulin sensitivity was impaired in HFF PEPCK1 ASO-treated rats. PEPCK1 ASO worsened hepatic steatosis, although without additional impairments in hepatic insulin signaling or activation of inflammatory signals in the liver. Instead, the development of hepatic insulin resistance and the decrease in hepatic glycogen synthesis during a hyperglycemic clamp was attributed to a decrease in hepatic glucokinase (GCK) expression and decreased synthesis of glycogen via the direct pathway. The decrease in GCK expression was associated with increased expression of activating transcription factor 3, a negative regulator of GCK transcription. These studies have demonstrated that PEPCK1 is integral to coordinating cellular metabolism in the liver and adipose tissue, although it does not directly effect hepatic glucose production or adipose glyceroneogenesis.


Assuntos
Adiposidade , Diabetes Mellitus Tipo 2/enzimologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Glicogênio Hepático/biossíntese , Fígado/metabolismo , Oligonucleotídeos Antissenso/genética , Fosfoenolpiruvato Carboxiquinase (GTP)/genética , Tecido Adiposo Branco/metabolismo , Animais , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/fisiopatologia , Dieta Hiperlipídica/efeitos adversos , Glucoquinase/genética , Glucoquinase/metabolismo , Humanos , Insulina/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Lipogênese , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosfoenolpiruvato Carboxiquinase (GTP)/metabolismo , Ratos , Ratos Sprague-Dawley
12.
Nat Commun ; 9(1): 3592, 2018 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-30181538

RESUMO

Adipocytes undergo pronounced changes in size and behavior to support diverse tissue functions, but the mechanisms that control these changes are not well understood. Mammary gland-associated white adipose tissue (mgWAT) regresses in support of milk fat production during lactation and expands during the subsequent involution of milk-producing epithelial cells, providing one of the most marked physiological examples of adipose growth. We examined cellular mechanisms and functional implications of adipocyte and lipid dynamics in the mouse mammary gland (MG). Using in vivo analysis of adipocyte precursors and genetic tracing of mature adipocytes, we find mature adipocyte hypertrophy to be a primary mechanism of mgWAT expansion during involution. Lipid tracking and lipidomics demonstrate that adipocytes fill with epithelial-derived milk lipid. Furthermore, ablation of mgWAT during involution reveals an essential role for adipocytes in milk trafficking from, and proper restructuring of, the mammary epithelium. This work advances our understanding of MG remodeling and tissue-specific roles for adipocytes.


Assuntos
Adipócitos/citologia , Metabolismo dos Lipídeos , Glândulas Mamárias Animais/citologia , Glândulas Mamárias Humanas/citologia , Adipócitos/metabolismo , Adipócitos Brancos/citologia , Adipócitos Brancos/fisiologia , Animais , Aleitamento Materno , Tamanho Celular , Células Epiteliais/citologia , Células Epiteliais/fisiologia , Ácidos Graxos/metabolismo , Feminino , Humanos , Lactação/fisiologia , Glândulas Mamárias Animais/fisiologia , Glândulas Mamárias Humanas/fisiologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Gravidez
13.
Development ; 145(17)2018 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-30045918

RESUMO

The embryonic origin of distinct fat depots and the role for ontogeny in specifying the functional differences among adipocyte lineages between and within depots is unclear. Using a Cre/Lox-based strategy to track the fate of major mesodermal subcompartments in mice we present evidence that <50% of interscapular brown adipocytes are derived from progenitors of the central dermomyotome. Furthermore, we demonstrate that depot-specific adipocyte lineages spatially diverge as early as gastrulation, and that perigonadal adipocytes arise from separate mesodermal subcompartments in males and females. Last, we show adipocyte precursors (APs) of distinct lineages within the same depot exhibit indistinguishable responses to a high fat diet, indicating that ontogenetic differences between APs do not necessarily correspond to functional differences in this context. Altogether, these findings shed light on adipose tissue patterning and suggest that the behavior of adipocyte lineage cells is not strictly determined by developmental history.


Assuntos
Adipócitos/metabolismo , Tecido Adiposo/embriologia , Linhagem da Célula/fisiologia , Embrião de Mamíferos/embriologia , Mesoderma/embriologia , Células-Tronco/metabolismo , Adipócitos/citologia , Tecido Adiposo/citologia , Animais , Embrião de Mamíferos/citologia , Mesoderma/citologia , Camundongos , Camundongos Transgênicos , Células-Tronco/citologia
14.
Cell Rep ; 22(8): 2133-2145, 2018 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-29466739

RESUMO

While therapeutic modulation of miRNAs provides a promising approach for numerous diseases, the promiscuous nature of miRNAs raises concern over detrimental off-target effects. miR-33 has emerged as a likely target for treatment of cardiovascular diseases. However, the deleterious effects of long-term anti-miR-33 therapies and predisposition of miR-33-/- mice to obesity and metabolic dysfunction exemplify the possible pitfalls of miRNA-based therapies. Our work provides an in-depth characterization of miR-33-/- mice and explores the mechanisms by which loss of miR-33 promotes insulin resistance in key metabolic tissues. Contrary to previous reports, our data do not support a direct role for SREBP-1-mediated lipid synthesis in promoting these effects. Alternatively, in adipose tissue of miR-33-/- mice, we observe increased pre-adipocyte proliferation, enhanced lipid uptake, and impaired lipolysis. Moreover, we demonstrate that the driving force behind these abnormalities is increased food intake, which can be prevented by pair feeding with wild-type animals.


Assuntos
Tecido Adiposo/patologia , Ingestão de Alimentos/genética , Deleção de Genes , Resistência à Insulina/genética , MicroRNAs/genética , Obesidade/genética , Tecido Adiposo/metabolismo , Adiposidade , Animais , HDL-Colesterol/sangue , HDL-Colesterol/metabolismo , LDL-Colesterol/sangue , Ativação Enzimática , Regulação da Expressão Gênica , Predisposição Genética para Doença , Células Germinativas/metabolismo , Mediadores da Inflamação/metabolismo , Metabolismo dos Lipídeos/genética , Fígado/metabolismo , Camundongos Endogâmicos C57BL , MicroRNAs/metabolismo , Modelos Biológicos , Obesidade/sangue , Obesidade/patologia , Proteína Quinase C-épsilon/metabolismo , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo
15.
Nature ; 550(7674): 119-123, 2017 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-28953873

RESUMO

Catecholamine-induced lipolysis, the first step in the generation of energy substrates by the hydrolysis of triglycerides, declines with age. The defect in the mobilization of free fatty acids in the elderly is accompanied by increased visceral adiposity, lower exercise capacity, failure to maintain core body temperature during cold stress, and reduced ability to survive starvation. Although catecholamine signalling in adipocytes is normal in the elderly, how lipolysis is impaired in ageing remains unknown. Here we show that adipose tissue macrophages regulate the age-related reduction in adipocyte lipolysis in mice by lowering the bioavailability of noradrenaline. Unexpectedly, unbiased whole-transcriptome analyses of adipose macrophages revealed that ageing upregulates genes that control catecholamine degradation in an NLRP3 inflammasome-dependent manner. Deletion of NLRP3 in ageing restored catecholamine-induced lipolysis by downregulating growth differentiation factor-3 (GDF3) and monoamine oxidase A (MAOA) that is known to degrade noradrenaline. Consistent with this, deletion of GDF3 in inflammasome-activated macrophages improved lipolysis by decreasing levels of MAOA and caspase-1. Furthermore, inhibition of MAOA reversed the age-related reduction in noradrenaline concentration in adipose tissue, and restored lipolysis with increased levels of the key lipolytic enzymes adipose triglyceride lipase (ATGL) and hormone sensitive lipase (HSL). Our study reveals that targeting neuro-immunometabolic signalling between the sympathetic nervous system and macrophages may offer new approaches to mitigate chronic inflammation-induced metabolic impairment and functional decline.


Assuntos
Adipócitos/metabolismo , Tecido Adiposo/metabolismo , Envelhecimento/metabolismo , Catecolaminas/metabolismo , Inflamassomos/metabolismo , Lipólise , Macrófagos/metabolismo , Tecido Adiposo/citologia , Tecido Adiposo/efeitos dos fármacos , Envelhecimento/efeitos dos fármacos , Envelhecimento/genética , Animais , Caspase 1/metabolismo , Catecolaminas/farmacologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Fator 3 de Diferenciação de Crescimento/deficiência , Fator 3 de Diferenciação de Crescimento/genética , Fator 3 de Diferenciação de Crescimento/metabolismo , Lipase/metabolismo , Lipólise/efeitos dos fármacos , Lipólise/genética , Camundongos , Monoaminoxidase/metabolismo , Inibidores da Monoaminoxidase/farmacologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/deficiência , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Norepinefrina/metabolismo , Esterol Esterase/metabolismo
16.
Adipocyte ; 6(3): 193-204, 2017 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-28872979

RESUMO

Adipocytes were identified in human bone marrow more than a century ago, yet until recently little has been known about their origin, development, function or interactions with other cells in the bone marrow. Little functional significance has been attributed to these cells, a paradigm that still persists today. However, we now know that marrow adipose tissue increases with age and in response to a variety of physiologic induction signals. Bone marrow adipocytes have recently been shown to influence other cell populations within the marrow and can affect whole body metabolism by the secretion of a defined set of adipokines. Recent research shows that marrow adipocytes are distinct from white, brown and beige adipocytes, indicating that the bone marrow is a distinct adipose depot. This review will highlight recent data regarding these areas and the interactions of marrow adipose tissue (MAT) with cells within and outside of the bone marrow.


Assuntos
Adipócitos/fisiologia , Tecido Adiposo/fisiologia , Células da Medula Óssea/citologia , Adipócitos/metabolismo , Adipócitos Marrons/metabolismo , Adipócitos Brancos/metabolismo , Adipogenia/fisiologia , Adipocinas/fisiologia , Tecido Adiposo Marrom/metabolismo , Tecido Adiposo Branco/metabolismo , Animais , Medula Óssea/metabolismo , Medula Óssea/fisiologia , Células da Medula Óssea/fisiologia , Diferenciação Celular/fisiologia , Humanos , Camundongos , Termogênese
17.
Adipocyte ; 6(3): 224-233, 2017 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-28792785

RESUMO

Over the past 2 decades, the incidence of childhood obesity has risen dramatically. This recent rise in childhood obesity is particularly concerning as adults who were obese during childhood develop type II diabetes that is intractable to current forms of treatment compared with individuals who develop obesity in adulthood. While the mechanisms responsible for the exacerbated diabetic phenotype associated with childhood obesity is not clear, it is well known that childhood is an important time period for the establishment of normal white adipose tissue in humans. This association suggests that exposure to obesogenic stimuli during adipose development may have detrimental effects on adipose function and metabolic homeostasis. In this study, we identify the period of development associated with puberty, postnatal days 18-34, as critical for the establishment of normal adipose mass in mice. Exposure of mice to high fat diet only during this time period results in metabolic dysfunction, increased leptin expression, and increased adipocyte size in adulthood in the absence of sustained increased fat mass or body weight. These findings indicate that exposure to obesogenic stimuli during critical developmental periods have prolonged effects on adipose tissue function that may contribute to the exacerbated metabolic dysfunctions associated with childhood obesity.


Assuntos
Tecido Adiposo Branco/metabolismo , Puberdade/fisiologia , Adipócitos/metabolismo , Tecido Adiposo/metabolismo , Tecido Adiposo Branco/fisiologia , Adiposidade/fisiologia , Animais , Diabetes Mellitus Tipo 2/etiologia , Diabetes Mellitus Tipo 2/metabolismo , Dieta Hiperlipídica/efeitos adversos , Feminino , Homeostase/fisiologia , Humanos , Leptina/metabolismo , Masculino , Camundongos , Obesidade/metabolismo , Puberdade/metabolismo
18.
Cell Metab ; 25(3): 493-494, 2017 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-28273471

RESUMO

Excess ECM and fibrosis of white adipose tissue (WAT) is associated with tissue dysfunction and type 2 diabetes. In this issue of Cell Metabolism, Marcelin et al. (2017) elucidate a key mechanism behind WAT fibrosis in which the activation of PDGFRα on adipocyte precursors drives this population toward a fibrotic phenotype.


Assuntos
Tecido Adiposo Branco/citologia , Diabetes Mellitus Tipo 2 , Adipócitos/citologia , Fibrose , Humanos , Fenótipo
19.
Cell Stem Cell ; 19(6): 738-751, 2016 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-27746098

RESUMO

Tissue growth and maintenance requires stem cell populations that self-renew, proliferate, and differentiate. Maintenance of white adipose tissue (WAT) requires the proliferation and differentiation of adipocyte stem cells (ASCs) to form postmitotic, lipid-filled mature adipocytes. Here we use the dynamic adipogenic program that occurs during hair growth to uncover an unrecognized regulator of ASC self-renewal and proliferation, PDGFA, which activates AKT signaling to drive and maintain the adipogenic program in the skin. Pdgfa expression is reduced in aged ASCs and is required for ASC proliferation and maintenance in the dermis, but not in other WATs. Our molecular and genetic studies uncover PI3K/AKT2 as a direct PDGFA target that is activated in ASCs during WAT hyperplasia and is functionally required for dermal ASC proliferation. Our data therefore reveal active mechanisms that regulate ASC self-renewal in the skin and show that distinct regulatory mechanisms operate in different WAT depots.


Assuntos
Adipócitos/citologia , Adipócitos/enzimologia , Autorrenovação Celular , Fator de Crescimento Derivado de Plaquetas/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Pele/citologia , Células-Tronco/citologia , Adipogenia , Animais , Antígeno CD24/metabolismo , Proliferação de Células , Derme/metabolismo , Perfilação da Expressão Gênica , Hiperplasia , Camundongos Endogâmicos C57BL , Modelos Biológicos , Fosfatidilinositol 3-Quinases/metabolismo , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo
20.
Cell Stem Cell ; 18(6): 684-686, 2016 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-27257753

RESUMO

Leptin is an adipocyte-derived hormone involved in energy sensing. In this issue of Cell Stem Cell, Yue et al. (2016) show that leptin is a physiologic signal that acts directly on Leptin-Receptor-expressing mesenchymal stromal cells in adult bone marrow to influence their lineage allocation in vivo, inhibiting bone formation and inducing marrow adipogenesis.


Assuntos
Medula Óssea/efeitos dos fármacos , Leptina , Adipócitos/efeitos dos fármacos , Adipogenia/efeitos dos fármacos , Células da Medula Óssea/efeitos dos fármacos , Osso e Ossos/efeitos dos fármacos , Células-Tronco Mesenquimais/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...